Fact-checked by Grok 2 weeks ago

New Drug Application

The New Drug Application (NDA) is the formal submission made by pharmaceutical sponsors to the (FDA) proposing approval for the marketing and distribution of a new drug product. Originating under the Federal Food, Drug, and Cosmetic Act of 1938, which mandated pre-market approval to prevent unsafe drugs from reaching consumers, the NDA serves as the cornerstone of U.S. drug regulation by requiring comprehensive evidence that the drug is safe and effective for its intended use. Sponsors must provide data from preclinical laboratory and animal studies, as well as human clinical trials conducted under an (IND) application, alongside details on drug composition, , manufacturing processes, and proposed labeling. Upon submission, the FDA's Center for Drug Evaluation and Research (CDER) conducts a multidisciplinary to evaluate whether the drug's benefits outweigh its known and potential risks, incorporating input from clinical, statistical, chemistry, manufacturing, and biopharmaceutics experts, and often advisory committees. Standard review timelines aim for completion within 10 months, while priority reviews for drugs addressing unmet needs in serious conditions target 6 months; expedited pathways such as Fast Track, , and Accelerated Approval further streamline development and assessment for qualifying products. Approval, if granted, authorizes interstate marketing, but includes post-approval requirements like risk evaluation and mitigation strategies (REMS) and confirmatory studies to monitor long-term safety and efficacy. This rigorous process, while ensuring protections, has evolved with user fee acts like the (PDUFA) to balance thoroughness with efficiency in bringing innovative therapies to market.

Historical Evolution

Origins and Early Legislation

The absence of federal drug regulation in the United States prior to the permitted widespread marketing of unsafe, adulterated, and falsely labeled pharmaceuticals, often derived from untested herbal or chemical sources, with no requirement for pre-market safety demonstrations. The , signed into law on June 30, 1906, established the initial federal framework by prohibiting the interstate shipment of adulterated or misbranded drugs and mandating disclosure of active ingredients on labels, while setting purity standards aligned with the and National Formulary. Enforcement relied on post-market inspections and seizures rather than prior approval, allowing novel drugs to reach consumers without evidence of safety, as long as labeling complied. This approach proved insufficient following the 1937 tragedy, in which the S.E. Massengill Company marketed an untested liquid formulation of the antibiotic dissolved in —a sweet but nephrotoxic solvent—resulting in approximately 107 deaths, primarily among children, and exposing the risks of absent pre-market scrutiny. In response, enacted the Federal Food, Drug, and Cosmetic Act on June 25, 1938, which amended the law to require manufacturers of new drugs to submit a New Drug Application (NDA) to the (FDA), including reports of investigations demonstrating through animal and human data, manufacturing details, and proposed labeling. The FDA was granted 60 days to review the application and raise objections; absent such action, the drug could proceed to market, marking the origin of the formal NDA process as a for pre-market clearance. This also authorized inspections and extended oversight to and medical devices, though proof remained unmandated until later amendments.

Major Amendments and Regulatory Shifts

The Kefauver-Harris Amendments, enacted on October 10, 1962, as part of the Federal Food, Drug, and Cosmetic Act, fundamentally altered the process by mandating proof of both safety and efficacy through "adequate and well-controlled investigations," shifting the evidentiary standard from manufacturer self-certification to explicit FDA pre-market approval. These changes, prompted by the thalidomide tragedy that caused thousands of birth defects in , also required in clinical trials and transferred regulatory authority over drug advertising from the to the FDA, thereby tightening oversight on promotional claims. The amendments extended average NDA review times from about 7 months pre-1962 to over 2 years by the mid-1960s, reflecting a deliberate prioritization of rigorous evidence over expediency. Subsequent regulatory shifts addressed approval delays amid growing drug development backlogs. The (PDUFA), signed into law on October 29, 1992, authorized the FDA to collect fees from pharmaceutical companies to fund additional review staff, reducing standard review timelines from approximately 30 months in the late 1980s to 10 months by the early 2000s without evidence of increased safety risks. PDUFA has been reauthorized five times (in 1997, 2002, 2007, 2012, and 2017), each iteration incorporating performance goals for review efficiency and introducing elements like priority review vouchers for in 2007. The Food and Drug Administration Modernization Act (FDAMA) of November 21, 1997, further streamlined NDA pathways by formalizing fast-track designations for serious conditions, expanding accelerated approval based on surrogate endpoints, and refining post-approval change reporting to balance manufacturing flexibility with . Later enactments, such as the of 2012, introduced designation for drugs showing substantial improvement over existing therapies, while the of December 13, 2016, permitted from sources like electronic health records to support approvals and expedited regenerative advanced therapies, aiming to incorporate patient-centered without lowering core thresholds. These shifts reflect an evolving emphasis on accelerating for unmet needs, with FDA indicating over 300 designations granted by 2023, though critics note reliance on surrogate endpoints may necessitate confirmatory post-market studies.

Pre-Market Development

Preclinical Testing Requirements

Preclinical testing, conducted prior to any human exposure, encompasses laboratory-based () and animal-based () studies designed to characterize a candidate's pharmacological activity, , and potential toxicity, thereby establishing a foundation for safe initiation of clinical trials. These studies must demonstrate that the investigational is reasonably safe for initial human use, as required under the Federal Food, , and Cosmetic Act and detailed in 21 CFR Part 312 for () applications, which precede New Application () submissions. The FDA mandates that preclinical data permit an adequate assessment of risks, including results from prior human experience (if any), animal , , and manufacturing details, with all nonclinical laboratory studies adhering to () regulations under 21 CFR Part 58 to ensure data integrity and reproducibility. Core requirements include developing a pharmacological profile to elucidate the drug's , receptor interactions, and dose-response relationships, often using models such as cell cultures or isolated tissues. assessments form the bulk of preclinical efforts, typically involving testing in at least two mammalian species (one and one non-rodent) to identify immediate adverse effects, followed by subchronic and chronic studies to detect delayed toxicities, (e.g., via or chromosomal aberration assays), carcinogenicity (in over 18-24 months), and reproductive/developmental toxicity in models like rats or rabbits. Pharmacokinetic studies evaluate absorption, distribution, metabolism, and excretion () to predict human dosing, often integrating metabolism data with animal models to inform safety margins, such as no-observed-adverse-effect levels (NOAELs) used to calculate starting doses for Phase I trials. The scope of testing is tailored to the drug's class, intended use, and prior data; for instance, biotechnology-derived pharmaceuticals may require species-specific assessments alongside standard , per FDA guidance. Range-finding studies precede definitive to refine doses, ensuring comprehensive coverage without unnecessary animal use, though the FDA has emphasized reducing reliance on animals where alternatives like modeling or human-relevant organoids show promise, as outlined in its 2025 roadmap. Failure to generate sufficient preclinical evidence of safety halts progression, as evidenced by rejections when data reveal unacceptable risks, underscoring the phase's role in filtering unsafe candidates early to minimize human harm.

Investigational New Drug Application

The application constitutes the initial regulatory submission to the U.S. by a sponsor—typically a pharmaceutical company or researcher—seeking authorization to initiate human clinical trials for an unapproved drug or biologic. This step follows preclinical testing, including animal , , and studies, to demonstrate that the drug is reasonably safe for initial human exposure and that the trial design is scientifically sound. Without an approved , interstate shipment of the investigational drug for clinical use is prohibited under the Federal Food, Drug, and Cosmetic Act. An IND submission must include comprehensive data across several modules, as outlined in 21 CFR Part 312. The introductory section covers general administrative information, such as the sponsor's details, name, and referenced prior submissions. , manufacturing, and controls () information details the drug substance's composition, manufacturing process, stability, and quality controls for the product and formulations. Nonclinical data encompasses , results from , and any prior human experience, justifying the proposed human dose and . The clinical protocol section provides the study design, including objectives, patient selection criteria, dosing regimen, monitoring plans, and statistical considerations for the proposed Phase 1, 2, or 3 trials. Additional requirements include investigator qualifications via Form FDA 1572, environmental assessments, and, for certain submissions, financial interest certifications on Form FDA 3454. Original IND applications are submitted electronically via the electronic Common Technical Document (eCTD) format or, in limited cases, in paper triplicate, accompanied by Forms FDA 1571 (Investigator's Brochure cover), 1572, and environmental impact details. The FDA assigns an application number upon receipt and conducts a 30-day review period to assess safety risks, potential clinical holds, and compliance with good clinical practices. If no clinical hold is imposed—grounds for which include insufficient nonclinical data, inadequate manufacturing controls, or unreasonable risk to subjects—trials may commence immediately after the 30-day window. Sponsors must submit annual reports, safety updates, and protocol amendments thereafter, with expedited reporting for serious adverse events within specified timelines, such as 7 days for life-threatening cases. INDs are categorized into commercial (for marketing intent), research (investigator-initiated for non-commercial studies), and treatment types, the latter allowing for patients without trial alternatives under compassionate use provisions. Emergency INDs enable immediate use in urgent situations, such as threats, bypassing standard timelines with verbal or written notifications followed by formal submission. These mechanisms, rooted in post-1938 regulations and refined by the 1962 Kefauver-Harris Amendments mandating efficacy data alongside safety, balance innovation with risk mitigation, though critics note that overly stringent preclinical thresholds can delay access to potentially beneficial therapies. Compliance failures may result in holds, termination, or enforcement actions, underscoring the application's role as a gatekeeper for ethical human testing.

Clinical Evaluation

Phase I Trials

Phase I trials represent the initial human testing phase following preclinical studies and approval of an (IND) application by the U.S. Food and Drug Administration (FDA). These trials primarily assess the safety, tolerability, and of the investigational drug in a small cohort, marking the transition from animal models to human subjects. The FDA requires sponsors to submit an IND at least 30 days prior to initiation, during which the agency reviews preclinical data to ensure no unreasonable risk to participants. Typically involving 20 to 100 participants, Phase I trials recruit healthy volunteers to minimize confounding factors from underlying diseases, though patients with the target condition—such as in trials—are used when ethical or practical constraints preclude healthy subjects due to potential . Trials last several months and employ dose-escalation designs, starting with low doses to identify the maximum tolerated dose (MTD) while monitoring for dose-limiting toxicities. Current Good Manufacturing Practice (CGMP) regulations under 21 CFR Part 211 apply, with tailored guidance for Phase I emphasizing in drug production to support safety data collection. Key objectives include evaluating pharmacokinetics—absorption, distribution, metabolism, and excretion—and pharmacodynamics, which measure the drug's biochemical and physiological effects, alongside preliminary efficacy signals if applicable. Safety endpoints dominate, focusing on adverse events, vital signs, electrocardiograms, and laboratory parameters under intensive monitoring, often in inpatient settings. Per 21 CFR 312.21, protocols must outline the investigation's scope, participant numbers, and risk mitigation, with the primary aim of confirming no serious harm before advancing to larger Phase II studies. Success rates remain low, with many drugs halted due to unacceptable toxicity or unfavorable profiles, underscoring the phase's role in early risk identification.

Phase II Trials

Phase II trials evaluate the preliminary of an investigational in patients with the target disease or condition, while continuing to monitor safety and optimal dosing. These studies typically involve administering the to a specified group of patients, often numbering from a few dozen to several hundred, to assess whether the treatment produces a therapeutic effect beyond what was observed in Phase I. The primary goal is to determine if the demonstrates sufficient promise to justify larger-scale Phase III testing, focusing on clinical endpoints such as symptom improvement or disease progression markers rather than solely . Trial designs in Phase II are frequently randomized and controlled, comparing the drug against or standard therapy to isolate its effects, though single-arm designs may be used for rare diseases or when ethical considerations preclude controls. Enrollment typically ranges from 100 to 300 patients, selected based on disease criteria outlined in the (IND) application, with protocols emphasizing statistical power to detect meaningful signals. Sponsors must submit detailed IND amendments for Phase II, including updated chemistry, manufacturing, and controls () information to ensure product consistency, as well as plans for monitoring adverse events and interim analyses. Duration varies from several months to two years, depending on the drug's mechanism and disease kinetics. Safety assessments remain integral, building on Phase I data to identify dose-limiting toxicities in a disease-specific , while is gauged through surrogate or clinical outcomes tailored to the therapeutic area. Regulatory guidance encourages adaptive designs in some cases, allowing modifications like dose adjustments based on interim data without compromising validity, though such adaptations require pre-approval to avoid bias. However, Phase II remains a high-attrition stage, with approximately 60-70% of drugs failing due primarily to insufficient (around 50% of failures), unexpected issues, or inadequate target engagement, underscoring the challenges in translating preclinical promise to . Successful Phase II outcomes inform Phase III planning, including endpoint selection and sample size calculations, but discrepancies between phases highlight risks such as over-optimistic effect sizes or population mismatches; FDA analyses of 22 cases revealed that even large Phase II trials assessing clinical outcomes failed to predict Phase III results in over half, often due to variability in patient heterogeneity or endpoint sensitivity. These trials thus serve as a critical gatekeeper, filtering candidates based on empirical evidence of biological activity while exposing causal gaps in drug-target hypotheses.

Phase III Trials

Phase III trials represent the confirmatory stage of clinical evaluation in drug development, designed to gather definitive evidence of a drug's efficacy and safety in large, diverse patient populations prior to submission of a New Drug Application (NDA) to the U.S. Food and Drug Administration (FDA). These trials typically involve randomized, controlled designs, often multicenter and double-blind, to minimize bias and establish whether the investigational drug provides a statistically significant treatment benefit over placebo, standard care, or active comparators. The primary objectives include verifying the drug's effectiveness for the intended indication, identifying rare adverse effects through extended exposure, and assessing benefits in subgroups such as varying ages, ethnicities, or comorbidities, thereby supporting the risk-benefit profile required for regulatory approval. Under FDA regulations, at least two adequate and well-controlled Phase III studies are generally required to demonstrate substantial evidence of effectiveness, as outlined in 21 CFR 314.126, with data from these trials forming the core of the clinical section in an NDA submission. Trial protocols emphasize rigorous statistical powering to detect clinically meaningful differences, with primary endpoints focused on objective measures like survival rates, symptom reduction, or changes, supplemented by secondary endpoints for broader outcomes. Enrollment typically ranges from hundreds to thousands of participants—often 300 to 3,000 or more per arm—to achieve sufficient statistical power (e.g., 80-90%) while ensuring generalizability across real-world demographics. These studies adhere to International Council for Harmonisation (ICH) guidelines and FDA's (IND) requirements for Phase 3, including detailed chemistry, manufacturing, and controls () information to ensure product consistency and quality during scaling. Duration varies by therapeutic area but averages a of 31 months, influenced by recruitment challenges, follow-up periods, and interim analyses for futility or efficacy. Costs for Phase III trials are substantial, reflecting the scale and complexity; a median expense per pivotal trial is approximately $19 million, though aggregate costs for multiple trials supporting an can exceed $100 million, driven by site management (14%), clinical procedures (20%), and staff (35% combined). Success rates from Phase III to approval hover between 60% and 73%, higher than earlier phases due to prior selection of promising candidates, though failures often stem from insufficient , unexpected signals, or inability to meet prespecified endpoints amid heterogeneous populations. Positive outcomes enable filing, where Phase III data must demonstrate the drug's benefits outweigh risks for the labeled population, potentially paving the way for full approval or accelerated pathways if endpoints are validated. Delays in these trials, common due to hurdles, can incur daily costs exceeding $55,000 in direct expenses for large studies.

NDA Submission and Review Process

NDA Contents and Filing

A New Drug Application (NDA) submitted to the U.S. Food and Drug Administration () must include specific components as outlined in 21 CFR 314.50 to enable evaluation of the drug's safety, efficacy, and manufacturing quality. The application begins with FDA Form 356h, which serves as the official transmittal form identifying the sponsor, drug name, and submission type. An organized index follows, providing a detailed table of contents for all sections, appendices, and references to facilitate FDA review. The core of the NDA comprises a summary and five to six technical sections. The summary, limited to no more than one-third of the total application length, integrates proposed labeling, foreign marketing history, technical summaries, and a benefit-risk assessment, emphasizing factual scientific data over promotional content. Technical sections detail: (1) chemistry, manufacturing, and controls (CMC), including drug substance and product composition, manufacturing processes, stability data, and quality controls; (2) nonclinical pharmacology and toxicology from animal studies; (3) human pharmacokinetics and bioavailability; (4) social, environmental, and abuse liability impacts (if applicable); (5) clinical data from controlled trials demonstrating safety and efficacy; and (6) statistical evaluations of study results. Supporting materials include case report tabulations summarizing patient data, full case report forms for pivotal studies, patent information, establishment descriptions for manufacturing sites, and debarment certifications ensuring sponsor eligibility. NDAs are filed electronically via the (eCTD) format to the FDA's Center for Drug Evaluation and Research (CDER), with submissions requiring prior pre-submission meetings recommended for complex applications. Concurrent payment of a (PDUFA) application fee is mandatory, categorized by whether clinical data are required: for fiscal year 2025, $4,310,002 for applications with clinical data and $2,155,001 without. Fees must be paid in U.S. currency via electronic methods such as debit or at submission to avoid refusal-to-file status. Upon receipt, FDA conducts a 60-day filing to assess ; if deficiencies exist, the application may be placed on hold or rejected, prompting resubmission.

FDA Evaluation and Timelines

Upon receipt of a New Drug Application (NDA), the U.S. (FDA) conducts an initial 60-day filing review to assess whether the submission is sufficiently complete for substantive evaluation; if deemed incomplete, the FDA issues a refuse-to-file (RTF) letter, halting the process until deficiencies are addressed. If filed, the application enters substantive review by a multidisciplinary team including physicians, statisticians, chemists, pharmacologists, and other experts who evaluate clinical data, , , manufacturing quality, and labeling. Under the Prescription Drug User Fee Act (PDUFA), which authorizes FDA to collect fees from drug sponsors to fund review operations, performance goals target completion of 90% of standard new molecular entity (NME) NDAs within 10 months of filing and 90% of priority NME NDAs within 6 months; these timelines represent goals rather than guarantees, with actual performance tracked annually. Priority review designation, granted within 60 days of NDA receipt for applications demonstrating potential significant improvement in safety or effectiveness over existing therapies, shortens the goal to 6 months, while standard review applies to other filings. Timelines may extend due to sponsor responses to FDA requests for additional data, major amendments, or advisory committee consultations, where external experts provide non-binding recommendations on complex or novel applications. The review culminates in an action letter—typically a complete response letter (CR) outlining deficiencies for resolution, or an approval letter if criteria are met—issued by the PDUFA goal date, though extensions via major amendment pauses can occur if sponsors submit substantial updates during review. Historical data indicate FDA meets or exceeds goals in most fiscal years, with enhancements from reauthorizations like (2023-2027) improving efficiency through process standardization and information technology. Delays beyond goals often stem from scientific complexities rather than regulatory hurdles, as evidenced by average review times for approved hovering around 8-9 months for priority cases in recent years.

Approval Types and Outcomes

The U.S. Food and Drug Administration (FDA) classifies reviews into standard and priority designations to allocate resources based on therapeutic potential. Standard review, the default pathway, targets a performance goal of 10 months for non-new molecular entity () applications and up to 12 months for under PDUFA goals, applying to drugs offering modest or no advancement over existing treatments. Priority review, granted upon request if the drug provides significant improvement in safety or effectiveness for a serious condition, shortens the goal to 6 months, facilitating faster access for therapies addressing unmet needs without compromising review rigor. These designations are determined early in the review process, typically within 60 days of receipt, and can apply to both traditional and expedited approval pathways. Traditional NDA approval requires substantial evidence of safety and effectiveness from adequate, well-controlled clinical investigations demonstrating direct clinical benefit, such as improved survival or reduced morbidity in the intended population. In contrast, accelerated approval, established under the 1992 regulations and codified in the FDA Safety and Innovation Act of 2012, permits approval for serious or life-threatening conditions with unmet medical needs based on endpoints or clinical endpoints reasonably likely to predict clinical benefit, rather than definitive outcomes. This pathway mandates confirmatory post-marketing studies to verify benefits, with the FDA empowered to withdraw approval or require label changes if trials fail to confirm efficacy; as of 2023, over 300 drugs have received accelerated approval, though some, like certain agents, have faced withdrawal for failing verification. can expedite either pathway, but accelerated approval emphasizes data to balance speed against requirements. NDA outcomes primarily include approval, issuance of a Complete Response Letter (CRL), or refusal to file. Approval authorizes marketing with a prescribing label detailing indications, dosing, and risks, often accompanied by boxed warnings or restricted distribution under Risk Evaluation and Mitigation Strategies (REMS) for high-risk drugs. A CRL, issued when an application is not approvable due to unresolved deficiencies in , clinical data, or safety, concludes the review cycle without final rejection, allowing sponsors to address issues and resubmit, potentially triggering a new review clock. Refusal to file occurs if the NDA lacks sufficient data for substantive review at filing, such as incomplete results, prompting refiling after supplementation. Withdrawals by sponsors or non-approvable resubmissions after CRLs represent effective denials, with FDA data indicating CRLs commonly cite bioanalytical, stability, or efficacy shortfalls in recent years.

Post-Approval Monitoring

Phase IV Studies

Phase IV studies, also known as studies, are conducted after a drug receives regulatory approval to monitor its long-term safety, efficacy, and usage patterns in broader, real-world populations that exceed the controlled settings of earlier clinical phases. These studies address limitations in pre-approval trials, such as smaller sample sizes that may miss rare adverse events occurring at rates below 1 in 1,000 to 1 in 10,000 exposures, and provide data on diverse patient subgroups, drug interactions, and off-label uses not fully evaluated prior to market entry. The U.S. (FDA) mandates or requests these studies under statutes like section 505(o) of the Federal Food, Drug, and Cosmetic Act, which authorizes requirements when postmarketing data reveal potential serious risks or when new evidence emerges post-approval. Postmarketing requirements (PMRs) are legally binding studies imposed by the FDA as a condition of approval or later, often to evaluate risks identified in approval data or , while postmarketing commitments (PMCs) are voluntary agreements by sponsors to conduct additional research, such as or comparative effectiveness studies. Sponsors must submit annual progress reports on PMRs and PMCs, detailing status, timelines, and any deviations, with FDA tracking compliance via internal databases updated as of August 9, 2024. IV encompasses a range of designs beyond traditional randomized controlled trials (RCTs), including observational cohort studies, pharmacoepidemiologic analyses, drug utilization reviews, and outcomes research on burden of illness or , all aimed at refining risk-benefit profiles. Findings from Phase IV studies can prompt regulatory actions, such as label updates to add warnings, contraindications, or dosing adjustments; for instance, expedited pathway drugs approved between 2005 and 2014 experienced safety-related label changes at a of 0.94 per drug per year, compared to 0.68 for standard approvals, highlighting how postmarketing data refines initial assessments. In cases of confirmed serious risks, these studies may lead to restricted distribution under Risk Evaluation and Mitigation Strategies (REMS), enhanced monitoring, or market withdrawal, ensuring ongoing causal evaluation of drug effects in uncontrolled settings where factors like and patient variability are prevalent. Non-compliance with PMRs can result in enforcement actions, including fines or approval revocation, underscoring their role in maintaining post-approval accountability.

Adverse Event Reporting and REMS

Post-approval adverse event reporting is mandated under sections 505 and 512 of the Federal Food, Drug, and Cosmetic Act, requiring drug sponsors to monitor and report safety data from commercial use to the FDA. Sponsors must submit periodic safety update reports and expedite reports of serious, unexpected adverse events within 15 calendar days of receipt, including follow-up information as it becomes available. These reports are primarily submitted electronically to the FDA Adverse Event Reporting System (FAERS), a database established in 2004 that aggregates millions of reports annually to detect safety signals, such as potential new risks not identified in pre-approval trials. As of November 2024, FAERS contains over 20 million reports, enabling FDA analyses for post-marketing safety surveillance, though reports are voluntary from consumers and healthcare providers and thus subject to underreporting and confounding factors like duplicate entries. The FDA uses FAERS data alongside other sources, including Phase IV studies and foreign regulatory reports, to identify signals of serious risks, which may prompt label updates, warnings, or market withdrawals. For instance, quarterly FAERS summaries highlight potential signals, with FDA applying disproportionality analyses like the proportional ratio to prioritize investigations. is enforced through inspections under the Postmarketing Adverse Event Compliance , which evaluates systems for timely detection and ; non-compliance can result in warning letters or enforcement actions. Healthcare professionals and patients can submit reports directly via MedWatch, but obligations remain primary for marketed products approved via . Risk Evaluation and Mitigation Strategies (REMS) complement adverse event reporting by imposing structured plans for drugs where serious risks could prevent widespread safe use, as authorized by the 2007 FDA Amendments Act. FDA requires REMS if labeling alone is insufficient to mitigate risks, with elements tailored to the drug's profile; as of May 2025, over 50 active REMS programs exist, covering drugs like opioids and . Core components include a risk mitigation goal, such as preventing fetal exposure or ensuring proper monitoring, implemented via medication guides, prescriber communications, or to Assure Safe Use (ETASU). ETASU may mandate prescriber certification, patient registries, or restricted distribution systems, with sponsors responsible for ongoing assessments of REMS effectiveness every 18 months or as triggered by new data. REMS integration with reporting allows FDA to refine strategies based on post-approval data; for example, if FAERS signals reveal inadequate risk mitigation, REMS modifications—such as adding ETASU—are mandated via supplemental applications. The REMS Public Dashboard, launched in 2024, provides on program statuses and assessments, aiding stakeholders in evaluating burden versus benefit. While REMS aim to balance access and safety, critiques note administrative burdens on prescribers and potential delays in care, though FDA data indicate they reduce targeted in high-risk populations. Sponsors face compliance inspections, with FDA able to impose civil penalties for failures impacting .

Criticisms and Controversies

Economic and Innovation Burdens

The New Drug Application (NDA) process imposes substantial economic burdens on pharmaceutical developers, primarily through the escalating costs of required clinical trials and . Estimates of the total capitalized cost to bring a single new drug to market range from $1 billion to over $2.6 billion, with much of this attributable to the large-scale III trials mandated for NDA submission to demonstrate and across diverse populations. These costs have risen sharply since the 1962 Kefauver-Harris Amendments, which elevated evidentiary standards, leading to trial sizes that can exceed thousands of participants and durations of several years per phase. Failure rates exacerbate the burden, as approximately 90% of drugs entering clinical development do not reach approval, amortizing losses across successful products and necessitating high pricing to recoup investments. The review timeline, while shortened by user fee acts like PDUFA to a of about 12.9 months for standard reviews, contributes to overall timelines averaging 10-15 years from , delaying revenue generation and increasing capital carrying costs estimated at hundreds of millions annually per project. This extended horizon heightens , particularly amid in R&D expenses outpacing general economic trends, with post-approval studies often adding further costs without guaranteed market expansion. Critics argue that such delays represent an implicit tax on , as foregone sales during approval can total billions for high-value therapeutics, compounded by competitive pressures from entry post-patent. These burdens stifle by prioritizing low-risk, incremental modifications over groundbreaking therapies, as evidenced by declining in areas like antibiotics where regulatory hurdles yield poor returns due to stewardship policies limiting use duration. Economic analyses indicate that stringent FDA requirements deter small firms, which lack the resources for protracted trials, fostering industry consolidation where large incumbents dominate 80-90% of approvals. Excessive regulation, per legal scholar , distorts toward compliance over novel science, reducing the pipeline of truly innovative drugs despite overall approval volumes.
AspectEstimated BurdenKey Driver
Capitalized R&D Cost per Approved Drug$1-2.6 billionMandated large-scale trials and high attrition
Total Development Timeline10-15 yearsSequential phases plus NDA review delays
Annual Delay Cost$100-500 million per projectLost revenue opportunity and financing expenses

Safety Failures and Over-Regulation Debates

The (NDA) process has been criticized for failing to detect certain safety risks prior to approval, leading to post-market withdrawals and significant impacts. For instance, (Vioxx), approved by the FDA in May 1999 for pain relief, was voluntarily withdrawn by Merck on September 30, 2004, after a revealed an approximate twofold increased risk of and with long-term use compared to . The drug had been prescribed to millions, with estimates linking it to around 27,785 heart attacks or sudden cardiac deaths in the U.S. alone. Early signals of cardiovascular risks emerged as early as 2000 from studies like VIGOR, but FDA advisory committees did not mandate stronger warnings until after further data accumulation, highlighting limitations in pre-approval trial durations (typically 3-6 months for low-risk populations) that may overlook rare or delayed adverse events. Similarly, extended-release oxycodone (OxyContin), approved in December 1995 for severe , contributed to the through inadequate scrutiny of potential and long-term claims during review. The FDA approved it without requiring studies supporting claims of reduced or suitability for non-cancer , despite limited on extended-release formulations; Purdue Pharma's amplified these issues, leading to widespread overprescription and . By 2017, opioid-related overdoses had claimed over 42,000 lives annually in the U.S., with prescription opioids like OxyContin implicated in initiating dependency pathways. Other examples include (Bextra), withdrawn in 2005 for cardiovascular and skin reaction risks after approval in 2001, and (Rezulin), pulled in 2000 for following 1997 approval. A 2017 analysis found that about one-third of drugs approved from 2001-2010 encountered post-approval safety warnings or withdrawals, often for cardiovascular or neurological issues not fully captured in III trials. These failures have fueled debates over whether the process is insufficiently rigorous or, conversely, overly burdensome, delaying beneficial therapies and inflating costs. Proponents of reform argue that stringent requirements—such as large-scale randomized controlled trials and extensive preclinical data—extend development timelines to 10-15 years and costs to $1-2.6 billion per approved drug, deterring and reducing the pipeline of new therapies. For example, comparisons with the () show U.S. review times averaging 306 days versus 383 days for in some datasets, but overall lags in U.S. approvals for certain drugs can reach 3-7 years due to FDA's higher evidentiary thresholds, potentially denying patients earlier access to treatments available in . Critics like those from the Manhattan Institute contend that such regulations prioritize theoretical over real-world benefits, stifling competition and contributing to fewer novel approvals annually (e.g., FDA approved 37 new molecular entities in versus historical peaks). Opposing views emphasize that post-approval withdrawals, while tragic, represent a small fraction—about 5% of approved drugs since 1950—of the thousands vetted, and attribute failures to industry influence rather than under-regulation, as seen in FDA advisory delays on Vioxx amid Merck's . Enhanced post-market tools like Phase IV studies and REMS have since addressed gaps, but debates persist on causal trade-offs: empirical evidence from device regulations suggests stricter FDA oversight correlates with safer products but fewer innovations and higher prices, mirroring drug concerns. Reforms proposed include adaptive trial designs and international harmonization to balance safety with efficiency, though entrenched bureaucratic incentives may perpetuate caution.

Industry Influence and Corruption Allegations

The pharmaceutical industry has been accused of exerting undue influence over the FDA's New Drug Application (NDA) review process through mechanisms such as the revolving door between agency regulators and industry positions, potentially compromising impartiality in drug approvals. Critics contend this dynamic fosters regulatory capture, where former FDA officials leverage insider knowledge to benefit sponsors, leading to higher approval rates for drugs from companies employing ex-regulators; one analysis found that hiring former FDA employees correlates with increased NDA success and elevated firm value. Such practices raise concerns about conflicts of interest, as evidenced by companies frequently recruiting FDA staffers who directly managed their successful NDA reviews. A prominent example of this involves FDA leadership: nine of the agency's past ten commissioners transitioned to roles in the or joined boards of drug companies post-tenure, including recent cases like the director of for Drug Evaluation and returning to in 2025. This pattern extends to rank-and-file reviewers, with data showing accelerated exits from the FDA amid turmoil, often to jobs that review handled by those same individuals during their tenure. Proponents of argue that such mobility undermines and prioritizes interests over rigorous safety evaluations in the NDA pathway. The Prescription Drug User Fee Act (PDUFA), enacted in 1992 and periodically reauthorized, amplifies these concerns by making the FDA financially dependent on industry payments, which constituted approximately 65% of the human drugs program's budget by the 2010s and fund NDA review timelines. Under PDUFA, drug sponsors pay fees to expedite reviews—reducing average NDA approval times from over two years pre-1992 to about ten months by 2017—but critics highlight a speed-safety tradeoff, with studies linking user-fee-driven deadlines to higher post-approval safety issues, including withdrawals and black-box warnings for drugs approved after PDUFA's implementation. This funding model has been described as institutional corruption, shifting regulatory priorities toward meeting industry timelines over independent patient-centered assessments. Allegations extend to broader systemic biases, such as suppressed adverse data during submissions or favoritism in accelerated approvals, though outright remains rare due to enforcement; the Department of Justice recovered $1.67 billion from pharmaceutical cases in FY2024, many involving falsified -related data. While some analyses reject claims of overt , emphasizing procedural flaws over intent, of industry sway—via expenditures exceeding $300 million annually and ex-officials' —suggests structural incentives that may erode the FDA's gatekeeping role in drug approvals. Reforms proposed include extended cooling-off periods for ex-regulators and reduced reliance on user fees to mitigate these risks.

Global and Comparative Perspectives

Equivalents in Other Jurisdictions

In the , the primary equivalent to the Food and Drug Administration's (FDA) New Drug Application (NDA) is the Marketing Authorisation Application (MAA), submitted to the (EMA) under the centralized procedure for novel medicinal products, which grants marketing authorization valid across all 27 EU member states plus , , and . The MAA dossier must include comprehensive modules on quality (chemical, pharmaceutical, and biological documentation), non-clinical safety data, clinical efficacy and safety trials, and plans, with validation occurring within 14 days of submission followed by an initial assessment phase. The Committee for Medicinal Products for Human Use (CHMP) conducts the scientific review, targeting 210 active assessment days, interrupted by clock-stops for applicant responses to queries, typically resulting in a positive opinion forwarded to the for final approval within 67 days. In , processes New Drug Submissions (NDS) for innovative pharmaceuticals and biologics containing new active substances, mirroring the by requiring of , , and from preclinical and clinical studies, along with details and labeling proposals. The review targets 300 calendar days for standard NDS, with priority review reduced to 180 days for drugs addressing unmet needs or serious conditions, involving multidisciplinary expert committees and potential site inspections. Notice of Compliance () issuance follows successful review, enabling market entry, while abbreviated pathways exist for subsequent entry biologics or extraordinary uses. Japan's (PMDA) reviews New Drug Applications (NDA) submitted for approval by the Ministry of Health, Labour and Welfare (MHLW), requiring Japanese-language dossiers with data on , , , clinical trials (often including Japan-specific bridging studies), and post-marketing commitments. Standard review timelines are 12 months from acceptance, shortened to 6-9 months for priority designation in cases of innovative therapies or drugs, with electronic data submission mandatory since to streamline processing. PMDA's expert consultations and foreign data extrapolation guidelines facilitate global alignment, though full approval hinges on MHLW endorsement. In , the () oversees registration of prescription medicines via an application process equivalent to the , encompassing , , , and bioequivalence data submitted through the electronic portal, with pathways tailored to novelty and risk. The pathway targets 200 working days for first-round evaluation, extendable for complex reviews or second-round data requests, while for high-need drugs aims for 150 days and provisional registration allows 2-year conditional approval pending confirmatory data. Registration on the Australian Register of Therapeutic Goods (ARTG) follows successful assessment, including GMP compliance verification.

Harmonization Efforts and Challenges

The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), established in 1990 through collaboration among regulatory authorities from the , , and the , along with representatives, has driven primary efforts to harmonize standards for new drug applications. Its guidelines address (Q series), (S series), (E series), and multidisciplinary topics (M series), enabling sponsors to compile a unified (CTD) dossier applicable across ICH regions, which was endorsed in 2000 and implemented starting in 2003. This framework has minimized redundant preclinical and clinical studies, with estimates indicating reductions in development timelines by up to 30% for multinational submissions and avoidance of duplicative equivalent to millions of procedures annually. Subsequent expansions of ICH membership—incorporating regulators from (2012), (2014), (2015), (2017), (2018), and others—have broadened to emerging markets, while initiatives like the 2024 reflection paper on real-world data for generating on medicine effectiveness aim to align post-approval requirements. For new drug applications, such as the U.S. FDA's and the EU's Authorisation Application (MAA), the CTD has standardized module structures, but targeted alignments continue in areas like testing and nonclinical study designs under guidelines finalized as early as 1994 for assessments. These efforts promote global access to innovative therapies, with ICH guidelines adopted or referenced by over 50 countries, facilitating faster regulatory reviews and reducing costs estimated at billions annually for developers. Challenges to full persist due to divergent national priorities, legal frameworks, and scientific interpretations, leading to incomplete alignment on specifics like chemistry, , and controls () data, where EU MAA dossiers often require more detailed environmental risk assessments than U.S. NDAs. Regulatory philosophies vary—e.g., Europe's precautionary approach versus the U.S. risk-benefit balancing—resulting in persistent differences in endpoints or post-approval commitments, with studies showing that even under ICH, approval timelines can differ by 6-12 months across regions. Political barriers, including data exclusivity periods (5 years in the U.S. versus 8+10 in the EU) and concerns, impede , while gaps in non-ICH countries exacerbate selective market entry by manufacturers facing inconsistent standards. These issues, compounded by varying enforcement capacities, underscore that while ICH has achieved partial technical alignment, systemic divergences rooted in policies and economic incentives limit broader uniformity.

References

  1. [1]
    New Drug Application (NDA) - FDA
    Jan 21, 2022 · For decades, the regulation and control of new drugs in the United States has been based on the New Drug Application (NDA).
  2. [2]
    Development & Approval Process | Drugs - FDA
    Aug 8, 2022 · These approaches, or designations, are meant to address specific needs, and a new drug application may receive more than one designation, if ...How Drugs are Developed · Novel Drug Approvals at FDA · Forms & Submission...
  3. [3]
    Milestones in US Food and Drug Law - FDA
    Jan 30, 2023 · Federal controls over the drug supply began with inspection of imported drugs in 1848, although the first federal biologics law, which addressed ...
  4. [4]
    Part I: The 1906 Food and Drugs Act and Its Enforcement | FDA
    Apr 24, 2019 · The basis of the law rested on the regulation of product labeling rather than pre-market approval. Drugs, defined in accordance with the ...
  5. [5]
    Summary of NDA Approvals & Receipts, 1938 to the present - FDA
    Jan 31, 2018 · The 1906 Food and Drugs Act prohibited the adulteration or misbranding of pharmaceuticals, but that law did not require premarket approval of drugs.
  6. [6]
    [PDF] A History of the FDA and Drug Regulation in the United States
    Jun 30, 2025 · Congress passes The Federal Food, Drug, and Cosmetic (FDC) Act of 1938, which requires that new drugs show safety before selling. This starts a ...
  7. [7]
    Federal Food, Drug, and Cosmetic Act (FD&C Act) - FDA
    Mar 29, 2018 · The Federal Food, Drug, and Cosmetic Act and subsequent amending statutes are codified into Title 21 Chapter 9 of the United States Code.Drugs and Devices · FD&C Act Chapter IV: Food · Short Title and Definitions
  8. [8]
    Reform, Regulation, and Pharmaceuticals — The Kefauver–Harris ...
    in the form of “adequate and well-controlled investigations”— before ...
  9. [9]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · "Also critically, the 1962 amendments required that the FDA specifically approve the marketing application before the drug could be marketed, ...Missing: impact | Show results with:impact
  10. [10]
    Food, Drug, and Cosmetic Act - StatPearls - NCBI Bookshelf
    The 1938 Federal Food, Drug, and Cosmetic Act (FDCA) is a set of United States (US) laws that authorize the Food and Drug Administration (FDA) to oversee and ...
  11. [11]
    Lost medicines: a longer view of the pharmaceutical industry with ...
    This project catalogues over-the-counter drugs and compounds lost following implementation of the 1962 Kefauver-Harris Amendment.
  12. [12]
    PDUFA Legislation and Background - FDA
    Dec 16, 2015 · The Prescription Drug User Fee Act (PDUFA) was created by Congress in 1992 and authorizes FDA to collect fees from companies that produce certain human drug ...
  13. [13]
    The Prescription Drug User Fee Act: Much More Than User Fees - NIH
    PDUFA legislation has instituted a broad range of changes to FDA policy, including evidentiary standards for drug approval, accelerated pathways for approval.
  14. [14]
    Food and Drug Administration Modernization Act (FDAMA) of 1997
    Mar 29, 2018 · The Food and Drug Administration Modernization Act (FDAMA), enacted Nov. 21, 1997, amended the Federal Food, Drug, and Cosmetic Act relating to the regulation ...Missing: NDA | Show results with:NDA
  15. [15]
    The Potential Effect of the 21st Century Cures Act on Drug ... - NIH
    The 21st Century Cures Act is a pivotal piece of legislation that will influence the FDA drug and device approval process for the foreseeable future.
  16. [16]
    Investigational New Drug (IND) Application - FDA
    Get to know the investigational new drug application (IND). This includes the types, laws and regulations, and emergency use of INDs.
  17. [17]
    21 CFR Part 312 -- Investigational New Drug Application - eCFR
    This part contains procedures and requirements governing the use of investigational new drugs, including procedures and requirements for the submission to, and ...Import and export requirements. · Title 21 · 312.20 – 312.38 · Administrative Actions
  18. [18]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · FDA requires researchers to use good laboratory practices (GLP), defined in medical product development regulations, for preclinical laboratory ...
  19. [19]
    Frequently Asked Questions on Drug Development and ... - FDA
    Aug 1, 2023 · What is an Investigational New Drug Application? In many ways, the investigational new drug (IND) application is the result of a successful ...
  20. [20]
    Preclinical Regulatory Requirements | Social Science Research ...
    Preclinical studies must follow FDA GLP regulations, including developing a pharmacological profile, determining acute toxicity in two animal species, and ...
  21. [21]
    Key IND-Enabling Studies Required for IND Approval | Allucent
    The key IND-enabling studies typically required include pharmacology, pharmacokinetics, and toxicology assessments.
  22. [22]
    [PDF] Guidance for Industry - S6 Preclinical Safety Evaluation of ... - FDA
    This FDA guidance provides principles for designing preclinical safety evaluation programs for biotechnology-derived pharmaceuticals, applicable to drug and ...
  23. [23]
    [PDF] Roadmap to Reducing Animal Testing in Preclinical Safety Studies
    Apr 10, 2025 · Current FDA requirements for mAbs mandate GLP- compliant repeat-dose toxicity studies (often 1–6 months duration) in animals, alongside ...<|separator|>
  24. [24]
    IND Applications for Clinical Investigations: Chemistry, Manufacturing
    Feb 25, 2022 · This component of an IND application includes the Chemistry, Manufacturing, and Control information for: (1) drug substance; (2) drug product; (3) placebo ...
  25. [25]
    Content and Format of Investigational New Drug Applications (INDs ...
    May 6, 2020 · This guidance document covers the content and format of Investigational New Drug Applications (INDs) for Phase 1 studies of drugs, including ...
  26. [26]
    [PDF] Investigational New Drug Applications (INDs) - FDA
    In general, the IND regulations in part 312 require that human research studies be conducted under an IND if all of the following conditions exist: Page 6 ...
  27. [27]
    IND Application Procedures: Overview - FDA
    Oct 9, 2015 · Original IND applications must be submitted in triplicate with forms 1571, 1572, and 3674. FDA will notify the sponsor of the application date. ...
  28. [28]
    Submission of an Investigational New Drug Application (IND) to CBER
    Sep 29, 2023 · IND Content and Format · Form FDA 1572: Statement of Investigator · Form FDA 3454: Certification: Financial Interests and Arrangements of Clinical ...
  29. [29]
    Safety Reporting Requirements for INDs and BA/BE Studies - FDA
    This document provides guidance to sponsors and investigators on expedited safety reporting requirements for human drug and biological products.<|separator|>
  30. [30]
    Investigational New Drug (IND) and Emergency ... - FDA
    Sep 16, 2025 · This table provides links to information for investigators about submitting Investigational New Drug (IND) and Emergency Investigational New ...
  31. [31]
    [PDF] FDA and Clinical Drug Trials: A Short History
    Initial regulations under the 1938 Act (issued December 28, 1938), required the person who introduced an investigational new drug into interstate commerce to ...
  32. [32]
    Step 3: Clinical Research - FDA
    Jan 4, 2018 · Phase 1 studies are closely monitored and gather information about how a drug interacts with the human body. Researchers adjust dosing schemes ...
  33. [33]
    FDA's Drug Review Process: Continued
    Aug 24, 2015 · Phase 1 studies (typically involve 20 to 80 people). Phase 2 studies (typically involve a few dozen to about 300 people). Phase 3 studies ( ...
  34. [34]
    Trial Phases 1, 2 & 3 Defined | Clinical Research Management (CRM)
    Phase I trials are concerned primarily with establishing a new drug's safety and dose range in about 20-100 healthy volunteers.
  35. [35]
    [PDF] Guidance for Industry CGMP for Phase 1 Investigational Drugs - FDA
    This FDA guidance provides CGMP recommendations for Phase 1 investigational drugs, including an introduction, background, scope, and general guidance.
  36. [36]
    [PDF] Considerations for the Design of Early-Phase Clinical Trials of ... - FDA
    For early-phase clinical trials, especially first-in-human trials, the primary objective should be an evaluation of safety (21 CFR 312.21).
  37. [37]
    An Overview of Phase II Clinical Trial Designs - PMC - NIH
    Phase II trials determine if a new treatment has promising efficacy and safety, usually with a few hundred patients, to warrant further phase III trials.
  38. [38]
    Phase II Trials in Drug Development and Adaptive Trial Design - PMC
    Jun 24, 2019 · Most phase II clinical studies have enrollments of 100 to 200 patients and multiple tests; therefore, the odds of multiple false positive ...
  39. [39]
    INDs for Phase 2 and Phase 3 Studies CMC Information - FDA
    Feb 22, 2018 · This guidance provides recommendations to sponsors of investigational new drug applications (INDs) on the chemistry, manufacturing, and controls (CMC) ...
  40. [40]
    [PDF] Adaptive Designs for Clinical Trials of Drugs and Biologics - FDA
    early-phase or exploratory clinical trials as well as trials conducted to satisfy post-marketing ... clinical trials, such as phase 1, phase 2, and phase. 3 (21 ...
  41. [41]
    Key indicators of phase transition for clinical trials through machine ...
    The most critical failures happen at later stages; ∼60–70% of Phase II trials and 30–40% of Phase III trials are unsuccessful; meaning that 60–70% of the drugs ...
  42. [42]
    Phase II and phase III failures: 2013–2015 | Nature Reviews Drug ...
    Nov 4, 2016 · a | The combined percentage of phase II and phase III clinical trial failures ... b | Reasons for failure in phase II clinical trials. c | Reasons ...
  43. [43]
    22 Case Studies Where Phase 2 and Phase 3 Trials Had Divergent ...
    Dec 21, 2017 · FDA studied 22 recent cases in which promising phase 2 clinical trial results were not confirmed in phase 3 clinical testing.
  44. [44]
    [PDF] 22 Case Studies Where Phase 2 and Phase 3 Trials Had ... - FDA
    These unexpected results could occur even when the phase 2 study was relatively large and even when the phase 2 trials assessed clinical outcomes. In two ...
  45. [45]
    21 CFR 314.50 -- Content and format of an NDA. - eCFR
    The NDA is required to contain tabulations of the data from each adequate and well-controlled study under § 314.126 (Phase 2 and Phase 3 studies as described ...
  46. [46]
    Design of Small Clinical Trials - NCBI - NIH
    Phase I trials are the earliest-stage clinical trials used to study an experimental drug in humans, are typically small (less than 100 participants), and are ...
  47. [47]
    Cost and Duration of Clinical Trials in Drug Development by ... - NIH
    Apr 11, 2025 · The median durations were 20 months for Phase 1, 29 months for Phase 2, and 31 months for Phase 3. The cost distributions were positively skewed ...
  48. [48]
    How much do phase III trials cost? | Nature Reviews Drug Discovery
    Oct 30, 2018 · The median expense for a single phase III trial is $19 million, they report in JAMA Internal Medicine, after assessing the details of 138 pivotal trials.
  49. [49]
    Inclusion of phase III clinical trial costs in health economic evaluations
    Oct 1, 2024 · The costs of phase III trials primarily involve administrative staff (20%), clinical procedures (20%), clinical staff (15%), site monitoring (14 ...
  50. [50]
    Probability of Success in Drug Development - Graaf - 2022
    Apr 19, 2022 · For example, between 2016 and 2020 the average industry success rates for phase II and phase III were 29–34% and 70–73%, respectively. Clinical ...
  51. [51]
    Benchmarking R&D success rates of leading pharmaceutical ...
    The success rates per phase were as follows: Phase I to Phase II at 64%; Phase II to Phase III at 32%; Phase III to new drug application (NDA) at 60%; and NDA ...
  52. [52]
    How Much Does a Day of Delay in a Clinical Trial Really Cost?
    Jun 6, 2024 · Phase III clinical trials had the highest direct cost per day at $55,716. Phase II clinical trials cost roughly half that amount at $23,737 ...
  53. [53]
    Prescription Drug User Fee Amendments - FDA
    Sep 30, 2025 · The Prescription Drug User Fee Act (PDUFA) was created by Congress in 1992 and authorizes FDA to collect user fees from persons that submit certain human drug ...
  54. [54]
    Step 4: FDA Drug Review
    Jan 4, 2018 · If it is not complete, the review team can refuse to file the NDA. If it is complete, the review team has 6 to 10 months to make a decision on ...
  55. [55]
    [PDF] PDUFA REAUTHORIZATION PERFORMANCE GOALS AND ... - FDA
    PDUFA reauthorization goals include reviewing 90% of standard NME NDA/BLA submissions within 10 months, and 90% of priority NME submissions within 6 months.
  56. [56]
    Priority Review - FDA
    Jan 4, 2018 · FDA informs the applicant of a Priority Review designation within 60 days of the receipt of the original BLA, NDA, or efficacy supplement.Fast Track, Breakthrough · Fast Track · Accelerated Approval
  57. [57]
    [PDF] NDA at the FDA
    NDA Review Timeline. Page 7. 7. Agenda. • Before NDA: Brief overview of the drug development. • NDA at FDA: terminology and timelines. • NDA at FDA: review ...
  58. [58]
    Prescription Drug User Fee Act (PDUFA) Performance Reports - FDA
    Sep 9, 2025 · Since the implementation of PDUFA I in 1993, FDA has used PDUFA resources to significantly reduce the time it takes to evaluate new drugs ...
  59. [59]
    Completed PDUFA VII Deliverables - FDA
    PDUFA must be reauthorized every five years, and in September 2022, the President signed into law the sixth reauthorization of PDUFA, which establishes the ...
  60. [60]
    [PDF] The New Drug Approval Process - FDA
    NDA Types: Priority vs Standard. • Standard Review. – Non- NME: 10-month clock. – NME (under the Program): 12-month clock. • Priority Review. – Non- NME: 6- ...
  61. [61]
    Accelerated Approval | FDA
    Feb 24, 2023 · These regulations allowed drugs for serious conditions that filled an unmet medical need to be approved based on a surrogate endpoint.
  62. [62]
    Accelerated Approval Program - FDA
    Aug 21, 2025 · The FDA instituted its Accelerated Approval Program to allow for earlier approval of drugs that treat serious conditions, and fill an unmet medical need based ...
  63. [63]
    Complete Response Letter Final Rule - FDA
    Feb 9, 2018 · We will send applicants a complete response letter to indicate that the review cycle for an application is complete and that the application is not ready for ...Missing: outcomes | Show results with:outcomes
  64. [64]
    Complete Response Letters (CRLs) - openFDA
    This database contains Complete Response Letters (CRLs) associated with approved and unapproved New Drug Applications (NDAs) and Biologics License Applications ...Missing: rejection | Show results with:rejection
  65. [65]
    Phase IV of Drug Development - PMC - NIH
    Not all Phase IV studies are post-marketing surveillance (PMS) studies but every PMS study is a phase IV study. Phase IV is also an important phase of drug ...
  66. [66]
    [PDF] Guidance for Industry: Postmarketing Studies and Clinical Trials - FDA
    Under section 505(o)(3)(D)(i), before requiring a postmarketing study, FDA must find that adverse event reporting under section 505(k)(1) of the Act and the ...
  67. [67]
    Postmarketing Requirements and Commitments: Introduction - FDA
    Aug 9, 2024 · Postmarketing requirements (PMRs) include studies and clinical trials that sponsors are required to conduct under one or more statutes or ...
  68. [68]
    Postmarketing Requirements and Commitments: Reports - FDA
    May 30, 2025 · This annual report is intended to summarize the data in the FDA internal PMR/PMC databases, which are used to track the status of PMRs and PMCs.Missing: Phase | Show results with:Phase
  69. [69]
    Safety related label changes for new drugs after approval in the US ...
    Sep 7, 2017 · Expedited pathway drugs were characterized by a rate of 0.94 safety related label changes for each drug per year, compared with 0.68 safety ...
  70. [70]
    FDA's Adverse Event Reporting System (FAERS)
    Nov 8, 2024 · The FDA Adverse Event Reporting System (FAERS) database contains adverse event reports, medication error reports and product quality complaintsFeedback form · FAQ · International Regulatory...
  71. [71]
    Postmarketing Adverse Event Reporting Compliance Program - FDA
    May 5, 2020 · FDA reviews postmarketing safety information and conducts postmarketing adverse event reporting inspections of companies to evaluate compliance with laws and ...
  72. [72]
    Postmarketing Safety Reporting for Human Drug and Biological ...
    Jan 29, 2020 · This guidance is intended to assist applicants and other responsible parties in fulfilling the FDA=s existing postmarketing safety reporting requirements.
  73. [73]
    Risk Evaluation and Mitigation Strategies | REMS - FDA
    May 20, 2025 · A Risk Evaluation and Mitigation Strategy (REMS) is a drug safety program that the US Food and Drug Administration (FDA) can require for certain medications ...What's in a REMS? · Public Dashboard · REMS Compliance Program
  74. [74]
    What's in a REMS? - FDA
    Jan 26, 2018 · REMS include a risk mitigation goal, and are comprised of information communicated to and/or required activities to be undertaken by one or more participants.
  75. [75]
    REMS Compliance Program - FDA
    Sep 22, 2022 · FDA conducts inspections to evaluate compliance with risk evaluation and mitigation strategies (REMS) requirements to ensure the drug's health benefits ...
  76. [76]
    Risk Evaluation and Mitigation Strategy (REMS) Public Dashboard
    Oct 2, 2024 · The REMS Public Dashboard is an interactive web-based tool that will allow for the analysis of REMS data in a user-friendly way.
  77. [77]
    Costs of Drug Development and Research and ... - JAMA Network
    Jun 28, 2024 · In this study, the mean cost of the clinical phase per drug candidate was estimated at $117.4 million, lower than the majority of the reported ...
  78. [78]
    Calling All Biopharmaceutical Companies: We Need ... - Tufts CSDD
    Joseph DiMasi, lead economist for the Tufts Center for the Study of Drug Development's (CSDD) recently released study results on the R&D costs of new drug ...
  79. [79]
    Costs of Drug Development and Research and Development ...
    Jun 28, 2024 · A 2012 study by Tufts Center for the Study of Drug Development estimated that nonclinical stage costs are substantially lower than the updated ...
  80. [80]
    Research and Development in the Pharmaceutical Industry
    Apr 8, 2021 · In recent studies, estimates of the average R&D cost per new drug range from less than $1 billion to more than $2 billion per drug. Those ...
  81. [81]
    Analysis of US Food and Drug Administration new drug and biologic ...
    Feb 9, 2024 · Over time, the median FDA review time significantly decreased from 26.6 (23.6) months in the pre-PDUFA period, to 12.9 (14.9) in the PDUDA- ...
  82. [82]
    Clinical development times for innovative drugs - PMC - NIH
    Receipt of a complete response letter from the FDA and other failures to win approval within the first review cycle increased review times by 829 days (95% CI ...
  83. [83]
    The economic challenges of new drug development - ScienceDirect
    We discuss key economic obstacles in the discovery of new drugs for infectious diseases, from novel antibiotics to diseases that primarily affect the poor.
  84. [84]
    [PDF] Stifling Innovation hearing v05 06 2024
    May 8, 2024 · FDA Reform to Promote Pharmaceutical Product Innovation for Small Companies. Regulatory barriers are magnified for entrepreneurs and small ...
  85. [85]
    Comparative analysis of FDA approvals by top 20 pharma ...
    FDA approved 176 new drugs for the 20 leading pharmaceutical companies (2014–2023). An additional 33 new drugs were acquired through post-approval acquisition.
  86. [86]
    Overdose How excessive government regulation stifles ... - NIH
    Dec 3, 2007 · In Overdose: how excessive government regulation stifles pharmaceutical innovation, Richard Epstein confronts the many problems besieging ...
  87. [87]
    New Estimates on the Cost of a Delay Day in Drug Development
    May 21, 2024 · In late 2023, the Tufts Center for the Study of Drug Development conducted empirical research to gather more accurate and granular estimates and ...
  88. [88]
    Rofecoxib (Vioxx) voluntarily withdrawn from market - PMC - NIH
    Oct 26, 2004 · Crawford, but “overall, patients taking the drug chronically face twice the risk of a heart attack compared to patients receiving a placebo.” ...Missing: timeline | Show results with:timeline
  89. [89]
    Vioxx (rofecoxib) Questions and Answers - FDA
    Apr 6, 2016 · The new study shows that Vioxx may cause an increased risk in cardiovascular events such as heart attack and strokes during chronic use. 7. What ...Missing: timeline | Show results with:timeline
  90. [90]
    FDA-Approved Prescription Drugs Later Pulled from the Market by ...
    Sep 20, 2025 · Vioxx (Rofecoxib) ... Cause for Recall: increased risk of heart attack and stroke; linked to about 27,785 heart attacks or sudden cardiac deaths ...
  91. [91]
    Failing the Public Health — Rofecoxib, Merck, and the FDA
    It was not until February 8, 2001, that the FDA Arthritis Advisory Committee met to discuss concern about the potential cardiovascular risks associated with ...Missing: timeline heart
  92. [92]
    [PDF] The sad story of Vioxx, and what we should learn from it
    Sep 30, 2004 · The approval was based on data from trials lasting 3 to 6 months and involving patients at low risk for cardiovascular illness.Missing: timeline | Show results with:timeline
  93. [93]
    How FDA Failures Contributed to the Opioid Crisis | Journal of Ethics
    The FDA did not properly enforce the Food, Drug, and Cosmetic Act when it approved Purdue Pharma's new drug application for extended-release (ER) oxycodone in ...
  94. [94]
    What led to the opioid crisis—and how to fix it | Harvard T.H. Chan ...
    Feb 9, 2022 · OxyContin approval is one example—Purdue Pharma was later shown to have presented a fraudulent description of the drug as less addictive than ...
  95. [95]
    FDA Requires Major Changes to Opioid Pain Medication Labeling to ...
    Jul 31, 2025 · Tragically, the new drug application for OxyContin was initially approved without study data supporting its long term use to treat pain in many ...
  96. [96]
    10 dangerous drugs recalled by the FDA - MDLinx
    Jul 24, 2019 · 10 dangerous drugs recalled by the FDA · Valdecoxib (Bextra). Time on the market: 2001-2005 · Pemoline (Cylert). Time on the market: 1975-2010.
  97. [97]
    One-Third Of New Drugs Had Safety Problems After FDA Approval
    May 9, 2017 · Bextra and Zelnorm were withdrawn because of cardiovascular risk, and Raptiva was withdrawn because of increased risk of a rare and fatal ...
  98. [98]
    High Drug Prices Don't Accelerate Innovation—Lower R&D Costs Do
    Apply Congressional scrutiny to FDA regulations that increase R&D costs by greater than $100 million. Historically, the FDA has paid no attention to the costs ...Missing: debates | Show results with:debates
  99. [99]
  100. [100]
    FDA approves drugs more quickly than peer agency in Europe
    Apr 5, 2017 · The median review time for FDA-approved drugs was quicker, 306 days compared to 383 days for EMA-approved drugs.
  101. [101]
    Drugs and Devices: Comparison of European and U.S. Approval ...
    Time lag between approval in Europe to approval in the United States (for those that were able to obtain FDA approval) was 3 to 7 years. Analysis of the quality ...
  102. [102]
    Innovation And Its Discontents: FDA Regulation, Competition, And ...
    Innovative drugs surely come with costs – at least in the short term – and the industry and government regulators should face concerns about drug pricing ...
  103. [103]
    How many FDA Approved drugs have been withdrawn and ... - CRIB
    Thalidomide (1957) - Withdrawn due to severe birth defects. 2. Phenylpropanolamine (2000) - Withdrawn due to increased risk of hemorrhagic stroke. 3. Rofecoxib ...
  104. [104]
    [PDF] Approval Costs and Innovation in Medical Technologies
    Sep 29, 2023 · I advance this debate by measuring the impact of FDA regulation on innovation, market structure, and safety in established medical device ...
  105. [105]
    Food and Drug Administration vs European Medicines Agency - NIH
    The median review time was longer at the EMA than FDA and was shorter for drugs undergoing FDA‐expedited programmes compared to the same drugs approved by the ...
  106. [106]
    Revolving doors: board memberships, hedge funds, and the FDA ...
    May 8, 2024 · Nine of the FDA's past 10 commissioners went on to work for the drug industry or serve on the board of directors of a drug company.
  107. [107]
    Unlocking the Revolving Door: How FDA-Firm Relationships Affect ...
    Dec 20, 2023 · I find that when pharmaceutical companies hire former FDA employees, the rate of drug approvals increases which in turn raises firm value.
  108. [108]
    FDA's revolving door: Companies often hire agency staffers who ...
    Jul 5, 2018 · FDA's revolving door: Companies often hire agency staffers who managed their successful drug reviews. Job changes raise conflict of interest ...
  109. [109]
    Top FDA Drug Regulator Returns to Pfizer: The Revolving Door ...
    Feb 24, 2025 · “Cavazonni's move demonstrates that the revolving door between the FDA and the industries it regulates is alive and well and continues to ...
  110. [110]
    With FDA in turmoil, the 'revolving door' with industry is spinning faster
    Apr 25, 2025 · Scores of FDA employees are searching for an exit from an agency in turmoil, particularly staff members tasked with reviewing drug applications.
  111. [111]
    A Look At How The Revolving Door Spins From FDA To Industry - NPR
    Sep 28, 2016 · A job reviewing drug applications at the Food and Drug Administration can be the springboard for a career in industry.
  112. [112]
    FDA's Revolving Door: Reckoning and Reform - Stanford Law School
    Feb 28, 2023 · The revolving door is one persistent source of undue influence that overshadows the discharge of FDA regulators' duties to the public.
  113. [113]
    Drug Money: FDA Depends on Industry Funding; Money Comes with…
    Dec 1, 2016 · Corruption & Abuse of Power · National Security · Our Work Show submenu ... The FDA user fee negotiations illustrate a broader phenomenon: the ...<|separator|>
  114. [114]
    The Prescription Drug User Fee Act: Cause for Concern? - PMC - NIH
    Feb 12, 2018 · Critics of PDUFA cite concerns including the potential for a lack of FDA independence and medication safety issues involving drugs approved after the existence ...
  115. [115]
    Drug-Review Deadlines and Safety Problems
    Mar 27, 2008 · Critics have argued that the user-fee program makes the agency too dependent on the industry it regulates and has led the FDA to focus ...
  116. [116]
    Institutional corruption of pharmaceuticals and the myth of safe and ...
    Meeting the needs of the drug companies has taken priority over meeting the needs of patients. Unless this corruption of regulatory intent is reversed, the ...
  117. [117]
  118. [118]
    CPG Sec. 120.100 Fraud, Untrue Statements of Material Facts, Bribery
    May 7, 2020 · ... drug companies were found to have falsified data submitted in premarket applications to FDA. ... fraud, untrue statements of material fact, ...Missing: allegations | Show results with:allegations
  119. [119]
    The FDA Drug Approval Process Has Problems—But It Is Not Corrupt
    Feb 6, 2023 · Is the US drug approval process corrupt? Many critics say that it is, leveling the charged rhetoric of corruption at the Food and Drug ...
  120. [120]
    The Revolving Door In Health Care Regulation
    Sep 5, 2023 · A revolving door between government and industry can render government agencies more vulnerable to regulatory capture.Abstract · Study Data And Methods · Study Results · Discussion
  121. [121]
    Marketing authorisation | European Medicines Agency (EMA)
    The European Medicines Agency (EMA) is responsible for the scientific evaluation of centralised marketing authorisation applications (MAA).Obtaining an EU marketing... · Pre-authorisation guidance · YahooMail
  122. [122]
    Obtaining an EU marketing authorisation, step-by-step
    Steps prior to submitting an application (updated) · Submission of eligibility request · Notification of intention to submit an application · Appointment of ...
  123. [123]
    Drug and health product submissions under review (SUR)
    The Submissions Under Review (SUR) Lists include new drug submissions containing new active substances (pharmaceuticals and biologics with an active ingredient ...New drug submissionsSupplemental submissions ...Generic submissions under ...Submission 'Class'
  124. [124]
    Guidance on management of drug submissions and applications
    Oct 1, 2025 · This guidance gives direction and guidance when managing information submitted in accordance with the Food and Drugs Act and its ...
  125. [125]
    Food and Drug Regulations ( CRC , c. 870) - Laws.justice.gc.ca
    1 (1) A manufacturer of a new drug may file an abbreviated new drug submission or an abbreviated extraordinary use new drug submission for the new drug where, ...<|separator|>
  126. [126]
    New Drug Review with Electronic Data
    From October 1, 2023 (application date), PMDA does not require to submit "Explanation of Electronic Study Data (Form A)" and "Explanation of Electronic Study ...
  127. [127]
    Frequently Asked Questions (FAQ) | Pharmaceuticals and Medical ...
    Q2-9. Can we file our application for product approval to PMDA in a language other than Japanese? ... No, PMDA does not accept applications in other languages.Pharmaceuticals and Medical... · Application for Product Approval · Reviews
  128. [128]
    Evolving Landscape of New Drug Approval in Japan and Lags ... - NIH
    For a new drug approval, the PMDA conducts a scientific review on the product application according to the legislations in Japan, and after PMDA approval, ...<|separator|>
  129. [129]
    Prescription medicines registration process
    The TGA registration process for prescription medicines has eight phases, including pre-submission, submission, and assessment, managed by milestones.Introduction · Overview · Pre-process activities · Registration process phases
  130. [130]
    Application process for prescription medicines
    To register a prescription medicine, a company must submit data, forms, and fees to the TGA. There are priority, provisional, and orphan drug pathways.
  131. [131]
    [PDF] Guidance for Industry - FDA
    By harmonizing the regulatory expectations in regions around the world, ICH guidelines have substantially reduced duplicative clinical studies, prevented ...Missing: NDA | Show results with:NDA
  132. [132]
    Introduction - International Regulatory Harmonization Amid ... - NCBI
    International regulatory harmonization aims to create globally harmonized, science-based standards for drug development, to improve efficiency and promote ...
  133. [133]
    Efficacy Guidelines - ICH
    The ICH Harmonised Guideline was finalised under Step 4 in March 1994. This document gives recommendations on the design and conduct of studies to assess the ...<|separator|>
  134. [134]
    [PDF] ich-reflection-paper-pursuing-opportunities-harmonisation-using ...
    Jul 25, 2024 · This paper discusses harmonization in using real-world data (RWD) to generate real-world evidence (RWE) for medicine effectiveness, addressing ...
  135. [135]
    International Regulatory Harmonization - FDA
    Jan 31, 2025 · Regulatory harmonization represents a process where regulatory authorities align technical requirements for the development and marketing of pharmaceutical ...
  136. [136]
    EU MAA vs. US NDA: Understanding Key Quality Differences
    Aug 1, 2024 · CMC differences between EU MAA & US NDA: steps for compliance & manufacturer considerations for a harmonised global dossier.
  137. [137]
    ICH Guidelines for Pharma: Overcoming Compliance Challenges
    Sep 26, 2024 · Challenges include complex guideline interpretation, adapting existing systems, and ensuring consistent application across different ...